Hostname: page-component-7bb8b95d7b-495rp Total loading time: 0 Render date: 2024-09-27T02:00:03.415Z Has data issue: false hasContentIssue false

The effects of conjugated linoleic acid supplementation on anthropometrics and body composition indices in adults: a systematic review and dose–response meta-analysis

Published online by Cambridge University Press:  06 September 2023

Omid Asbaghi
Affiliation:
Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
Ghazaleh Shimi
Affiliation:
Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
Fatemeh Hosseini Oskouie
Affiliation:
Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
Kaveh Naseri
Affiliation:
School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
Reza Bagheri
Affiliation:
Department of Exercise Physiology, University of Isfahan, Isfahan, Iran
Damoon Ashtary-Larky
Affiliation:
Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
Michael Nordvall
Affiliation:
Department of Health and Human Performance, Marymount University, Arlington, VA, USA
Samira Rastgoo
Affiliation:
Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
Mohammad Zamani*
Affiliation:
Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
Alexei Wong
Affiliation:
Department of Health and Human Performance, Marymount University, Arlington, VA, USA
*
*Corresponding author: Dr M. Zamani, email md_zamany@yahoo.com
Rights & Permissions [Opens in a new window]

Abstract

Prior meta-analytic investigations over a decade ago rather inconclusively indicated that conjugated linoleic acid (CLA) supplementation could improve anthropometric and body composition indices in the general adult population. More recent investigations have emerged, and an up-to-date systematic review and meta-analysis on this topic must be improved. Therefore, this investigation provides a comprehensive systematic review and meta-analysis of randomised controlled trials (RCT) on the impact of CLA supplementation on anthropometric and body composition (body mass (BM), BMI, waist circumference (WC), fat mass (FM), body fat percentage (BFP) and fat-free mass (FFM)) markers in adults. Online databases search, including PubMed, Scopus, the Cochrane Library and Web of Science up to March 2022, were utilised to retrieve RCT examining the effect of CLA supplementation on anthropometric and body composition markers in adults. Meta-analysis was carried out using a random-effects model. The I2 index was used as an index of statistical heterogeneity of RCT. Among the initial 8351 studies identified from electronic databases search, seventy RCT with ninety-six effect sizes involving 4159 participants were included for data analyses. The results of random-effects modelling demonstrated that CLA supplementation significantly reduced BM (weighted mean difference (WMD): −0·35, 95 % CI (−0·54, −0·15), P < 0·001), BMI (WMD: −0·15, 95 % CI (−0·24, −0·06), P = 0·001), WC (WMD: −0·62, 95% CI (−1·04, −0·20), P = 0·004), FM (WMD: −0·44, 95 % CI (−0·66, −0·23), P < 0·001), BFP (WMD: −0·77 %, 95 % CI (−1·09, −0·45), P < 0·001) and increased FFM (WMD: 0·27, 95 % CI (0·09, 0·45), P = 0·003). The high-quality subgroup showed that CLA supplementation fails to change FM and BFP. However, according to high-quality studies, CLA intake resulted in small but significant increases in FFM and decreases in BM and BMI. This meta-analysis study suggests that CLA supplementation may result in a small but significant improvement in anthropometric and body composition markers in an adult population. However, data from high-quality studies failed to show CLA’s body fat-lowering properties. Moreover, it should be noted that the weight-loss properties of CLA were small and may not reach clinical importance.

Type
Systematic Review and Meta-Analysis
Copyright
© The Author(s), 2023. Published by Cambridge University Press on behalf of The Nutrition Society

Obesity negatively influences overall health, adds significantly to societal and economic burdens, and shows no signs of slowing(Reference Orukwowu1). Globally, millions of individuals maintain a sedentary lifestyle and adhere to nutrient-poor and energy-dense diets, which contributes to overweightness/obesity and increases the risk for many non-communicable chronic diseases(Reference Hutchesson, Gough and Müller2,Reference Wang, McPherson and Marsh3) . Therefore, the identification of alternative adiposity-reducing strategies with the potential to prevent or alleviate the negative consequences of obesity is warranted. Apart from lifestyle modification, often considered the cornerstone of a weight management programme(Reference Ashtary-Larky, Bagheri and Bavi4Reference Ashtary-Larky, Bagheri and Abbasnezhad6), a wide range of supplements are now available touting anti-obesity properties(Reference Asbaghi, Naeini and Ashtary-Larky7Reference Bagheri, Negaresh and Motevalli10). Among those, conjugated linoleic acid (CLA) has shown promise as a food supplement to reduce adiposity in preventing overweightness and obesity(Reference Chang, Gan and Liao11,Reference Mądry, Malesza and Subramaniapillai12) .

CLA have been investigated for various beneficial effects, including cancer, atherosclerosis and obesity(Reference Basak and Duttaroy13Reference O’Reilly, Lenighan and Dillon16). Major isomers of CLA are cis-9, trans-11 CLA (9, 11 CLA) and trans-10, cis-12 CLA (10, 12 CLA)(Reference Asbaghi, Ashtary-Larky and Naseri17), which are found naturally in ruminant animal food products(Reference Haghighat, Shimi and Shiraseb18) and are primary components of widely consumed CLA weight-loss supplements(Reference Ashwell, Ceddia and House19,Reference Churruca, Fernández-Quintela and Zabala20) . Although humans can produce endogenous CLA, the blood and tissue levels of CLA in non-supplemented individuals are less(Reference Sato, Shinohara and Honma21,Reference Turpeinen, Mutanen and Aro22) . According to prior investigations, isomer 10, 12 CLA seems to elicit the greatest beneficial effect on promoting weight loss in animals and humans(Reference Chen, Lin and Huang23Reference Mądry, Chudzicka-Strugała and Grabańska-Martyńska25). One proposed explanation behind CLA action may be stimulating apoptotic mechanisms and regulating lipolytic pathways, both of which positively affect body composition and weight loss in humans(Reference Baddini Feitoza, Fernandes Pereira and Ferreira da Costa26). A substantial body of evidence also indicates that CLA promotes weight loss by reducing fat cells’ size and altering fat cells’ evolution(Reference Brown and McIntosh27). While future research needs to elucidate further the physiological or other mechanisms behind CLA-induced altered fat cells, the vast majority of literature on the role of CLA in managing obesity utilises common measures for anthropometrics and body composition, including body mass (BM), BMI, waist circumference (WC) and body fat percentage (BFP)(Reference Joseph, Wasir and Misra28,Reference Pi-Sunyer29) . To this, a series of recent well-controlled pharmacological investigations have demonstrated conflicting results on the effectiveness of CLA supplementation on these outcomes in adults(Reference Chang, Gan and Liao11,Reference Abedi, Aref-Hosseini and Khoshbaten30) .

As noted, investigations on the association between CLA supplementation with anthropometric and body composition outcomes have sometimes been in agreement, which may be due to various factors, including supplementation dosages, the length of intervention and the health status of participants. Although prior meta-analyses exist, such investigations targeted special populations, including overweight and obese individuals(Reference Namazi, Irandoost and Larijani31,Reference Onakpoya, Posadzki and Watson32) and those with metabolic syndrome(Reference Kim, Lim and Lee33). To the best of our knowledge, two prior meta-analyses have been conducted to determine the pooled effects of CLA supplementation on fat and fat-free mass (FFM) in the general adult population(Reference Whigham, Watras and Schoeller34,Reference Schoeller, Watras and Whigham35) . However, these meta-analytic investigations, in particular, were performed over a decade ago, and numerous relevant randomised controlled trials (RCT) have been published since. Therefore, we performed a comprehensive systematic review and meta-analysis of the literature to date on the effects of CLA supplementation on anthropometric and body composition markers in adults.

Methods

This investigation was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses protocol(Reference Moher, Shamseer and Clarke36).

Search strategy

A comprehensive literature search was performed for RCT that investigated the efficacy of CLA supplementation on anthropometric measurements and body composition indicators using online databases, including PubMed/MEDLINE, Scopus, Web of Science, and Cochrane Library up to March 2022. The following MeSH and non-MeSH terms were applied in the search strategy: (‘Conjugated linoleic acid’ OR ‘conjugated fatty acid’ OR ‘bovic acid’ OR ‘rumenic acid’ OR ‘CLA’ AND Intervention OR ‘Intervention Study’ OR ‘Intervention Studies’ OR ‘controlled trial’ OR ‘randomized’ OR ‘randomized’ OR ‘random’ OR ‘randomly’ OR ‘placebo’ OR ‘clinical trial’ OR ‘Trial’ OR ‘randomized controlled trial’ OR ‘randomized clinical trial’ OR ‘RCT’ OR ‘blinded’ OR ‘double blind’ OR ‘double blinded’ OR ‘trial’ OR ‘clinical trial’ OR ‘trials’ OR ‘Pragmatic Clinical Trial’ OR ‘Cross-Over Studies’ OR ‘Cross-Over’ OR ‘Cross-Over Study’ OR ‘parallel’ OR ‘parallel study’ OR ‘parallel trial’).

No restrictions were placed in database searches for the date of publication. Reference lists of all relevant studies were cross-checked against database search results for overlooked publications. All references were included in the Endnote software (EndNote X21, Thomson Reuters, New York) for screening, and duplicate citations and unpublished manuscripts were removed.

Study selection and eligibility criteria

Titles and abstracts of all records from the initial search were evaluated independently by two investigators. Studies were selected for further analysis if they met the following criteria: (a) original RCT with either parallel or crossover designs; (b) studies that were done on adult participants (≥18 years old); (c) trials investigating the impact of CLA supplementation on anthropometric measurements (BM, BMI and WC) and body composition indicators (fat mass (FM), BFP and FFM) in both intervention and placebo groups; (d) studies that reported means and standard deviations for each outcome or any other effect sizes by which the calculation of means and standard deviation was possible. Conversely, studies were excluded if: (a) the duration of intervention was less than 4 weeks; (b) inadequate data on the selected outcomes in intervention or control groups was presented; (c) they were observational, case reports, reviews, letters to an editor, editorial articles, and in vitro studies and animal experiments; (d) children or adolescents were enrolled; and (e) no control group was apparent.

Data extraction

The data extraction was independently performed using a pre-designed standardised electronic form (Excel, Microsoft Office). The following information from each study were extracted: first author’s name, year of publication, study location, total sample size, numbers of cases (those who received CLA) and controls, participant’s demographic data (sex, mean age and BMI), the health status of participants, study design, the intervention dose, length of follow-up, and outcomes measured as mean and standard deviation of selected end points at study baseline, post-intervention, and/or changes between baseline and post-intervention.

Quality and certainty assessment

A systematic bias assessment of the included studies was performed using the Cochrane criteria(Reference Higgins, Thomas and Chandler37). The quality of all eligible studies was evaluated based on the following items: random sequence generation, allocation concealment, reporting bias, performance bias, detection bias, attrition bias and other possible causes of bias. Based on the Cochrane Handbook recommendation, studies were ranked as low (L), high risk of bias (H) or unclear (U) regarding each field of bias(Reference Higgins, Thomas and Chandler37). In addition, the overall certainty of evidence across the studies was evaluated based on the GRADE (Grading of Recommendations Assessment, Development, and Evaluation) Working Group guidelines. Subsequently, the quality of evidence was classified into four categories: high, moderate, low and very low(Reference Guyatt, Oxman and Vist38).

Data synthesis and statistical analysis

Data analysis was performed using STATA® version 14.0 (StataCorp.), and mean (sd) changes of outcomes were used to estimate the overall effect size. Effect sizes for all variables are reported as weighted mean differences (WMD) and 95 % CI derived from random-effects models. A random-effects model was selected to address significant heterogeneity between studies for methodology, outcome measures and participant characteristics. The I 2 index was used as an index of statistical heterogeneity of RCT. If the sd change following intervention was not reported in studies, it was calculated based on the formula provided by the Cochrane Collaboration(Reference Higgins39) such that: sd = square root [(sd baseline) 2 + (sd final) 2 – (2R × sd baseline × sd final)], where the correlation coefficient (R) = 0·8. Statistical heterogeneity between studies was assessed by Cochrane’s Q test (significance point at P < 0·1) and the I 2 index (significance point at I 2 > 40 %).

The publication bias was assessed using visual inspection of funnel plots and statistically using Egger’s regression and Begg’s tests(Reference Egger, Davey Smith and Schneider40). Subgroup analyses were performed to find probable sources of heterogeneity based on several predefined variables, including duration of follow-up (≥12 v. <12 weeks), intervention dosage (≥3 v. <3 g/d), participants’ health condition (healthy v. unhealthy), baseline values of BMI (normal v. overweight v. and obese), sex (female v. male v. combined) and the quality of studies (high v. moderate, v. high quality). Sensitivity analysis was applied to detect if an overall effect size relied on the outcomes of a particular RCT. To determine the non-linear dose–response and linear meta-regression effects of CLA dosage (g/d) as well as the duration of intervention on each marker, fractional polynomial modelling was used. A P-value 0·05 was considered to be a statistically significant outcome.

Results

Study selection

The study selection process is illustrated in Fig. 1. A total of 8351 publications were found in the initial search. Of those, 2419 were duplicates and thus removed from further consideration. After a review of the remaining 5932 titles and abstracts, 104 publications were advanced for full-text examination. An additional thirty-four studies were removed following full-text scrutiny according to inclusion/exclusion criteria. Finally, sixty-nine RCT with ninety-five effect sizes and 4159 participants met the inclusion criteria for quantitative and qualitative analyses.

Fig. 1. Flow chart of study selection for inclusion trials in the systematic review.

Study characteristics

The characteristics of the included RCT are outlined in Table 1. Investigations were published between 2000 and 2020 and were carried out in Europe(Reference Mądry, Chudzicka-Strugała and Grabańska-Martyńska25,Reference Berven, Bye and Hals41Reference Thom, Wadstein and Gudmundsen68) , Asia(Reference Chen, Lin and Huang23,Reference Abedi, Aref-Hosseini and Khoshbaten30,Reference Aryaeian, Shahram and Djalali69Reference Zh, Rastmanesh and Hehayati87) , America(Reference Adams, Hsueh and Alford88Reference Plourde103), Africa(Reference Goedecke, Rae and Smuts104Reference Lambert, Goedecke and Bluett106) and Oceania(Reference Attar-Bashi, Weisinger and Begg107). Of the sixty-nine RCT, six were randomised crossover design(Reference Rubin, Herrmann and Much63,Reference Sneddon, Tsofliou and Fyfe65,Reference Desroches, Chouinard and Galibois92,Reference Joseph, Jacques and Plourde93,Reference Venkatramanan, Joseph and Chouinard98,Reference Plourde103) , and the remaining were of parallel design. Intervention dosages of CLA varied between 1·0 and 6·8 g/d, and follow-up durations ranged from 4 to 104 weeks. Selected studies enrolled participants with different health conditions; three studies enrolled patients with diabetes(Reference Norris, Collene and Asp55,Reference Shadman, Taleban and Saadat78,Reference Zh, Rastmanesh and Hehayati87) ; three investigated the effects of CLA supplementation in individuals with hyperlipidemia(Reference Joseph, Jacques and Plourde93,Reference Venkatramanan, Joseph and Chouinard98,Reference Plourde103) , two recruited patients with metabolic syndrome(Reference Risérus, Arner and Brismar59,Reference Carvalho, Uehara and Rosa90) , and individual studies investigated the following health conditions: rheumatoid arthritis(Reference Aryaeian, Shahram and Djalali69), hypertension(Reference Zhao, Zhai and Wang80), atherosclerosis(Reference Hassan Eftekhari, Aliasghari and Babaei-Beigi76), chronic obstetric pulmonary disease(Reference Ghobadi, Matin and Nemati75) and benign breast disease(Reference Rezvani, Montazeri and Baradaran84). All remaining studies were performed on apparently healthy individuals. The vast majority of RCT were performed on both sexes, except sixteen investigations that were conducted exclusively on females(Reference Mądry, Chudzicka-Strugała and Grabańska-Martyńska25,Reference Mądry, Malesza and Subramaniapillai50,Reference Norris, Collene and Asp55,Reference Colakoglu, Colakoglu and Taneli72,Reference Kim, Kim and Ha82,Reference Rezvani, Montazeri and Baradaran84Reference Tavakkoli Darestani, Hosseinpanah and Tahbaz86,Reference Brown, Trenkle and Beitz89,Reference Carvalho, Uehara and Rosa90,Reference Medina, Horn and Keim95,Reference Ribeiro, Pina and Dodero97,Reference Zambell, Keim and Van Loan100Reference Pina, Ribeiro and Dodero102,Reference Lambert, Goedecke and Bluett106) and seventeen on males(Reference Pfeuffer, Fielitz and Laue57Reference Rubin, Herrmann and Much63,Reference Sneddon, Tsofliou and Fyfe65,Reference Bulut, Bodur and Colak70,Reference Tajmanesh, Aryaeian and Hosseini79,Reference Adams, Hsueh and Alford88,Reference Deguire, Makarem and Vanstone91Reference Joseph, Jacques and Plourde93,Reference Plourde103,Reference Kreider, Ferreira and Greenwood105,Reference Lambert, Goedecke and Bluett106) . The mean age of individuals was between 18 and 63·3 years, with BMI ranging from 19 to 37·1 kg/m2. All sixty-nine included trials had an appropriately controlled design, with the sole difference between control and treatment groups being the CLA intervention.

Table 1. Characteristics of the included studies

R, randomised; PC, placebo-control; DB, double-blind; M, male; F, female.

Risk of bias assessment

Cochrane risk of bias results of included studies is shown in Supplementary 1 and indicated that thirty-three(Reference Mądry, Chudzicka-Strugała and Grabańska-Martyńska25,Reference Abedi, Aref-Hosseini and Khoshbaten30,Reference Berven, Bye and Hals41,Reference Gaullier, Halse and Høye44,Reference Kamphuis, Lejeune and Saris46,Reference Kamphuis, Lejeune and Saris47,Reference Malpuech-Brugère, Verboeket-Van de Venne and Mensink51,Reference Nazare, de la Perrière and Bonnet53,Reference Pfeuffer, Fielitz and Laue57,Reference Risérus, Arner and Brismar59Reference Risérus, Vessby and Arnlöv62,Reference Sneddon, Tsofliou and Fyfe65Reference Bulut, Bodur and Colak70,Reference Colakoglu, Colakoglu and Taneli72,Reference Zhao, Zhai and Wang80,Reference Fouladi, Peng and Mohaghehgi81,Reference Brown, Trenkle and Beitz89Reference Joseph, Jacques and Plourde93,Reference Medina, Horn and Keim95,Reference Venkatramanan, Joseph and Chouinard98Reference Zambell, Keim and Van Loan100,Reference Plourde103,Reference Lambert, Goedecke and Bluett106) trials were considered to be at high risk for bias, nineteen(Reference Blankson, Stakkestad and Fagertun42,Reference Gaullier, Halse and Høivik43,Reference Mądry, Malesza and Subramaniapillai50,Reference Mougios, Matsakas and Petridou52,Reference Noone, Roche and Nugent54,Reference Nugent, Roche and Noone56,Reference Raff, Tholstrup and Basu58,Reference Rubin, Herrmann and Much63,Reference Hassan Eftekhari, Aliasghari and Babaei-Beigi76Reference Tajmanesh, Aryaeian and Hosseini79,Reference Kim, Kim and Ha82,Reference Park, Kim and Kim83,Reference MacRedmond, Singhera and Attridge94,Reference Pinkoski, Chilibeck and Candow96,Reference Pina, Ribeiro and Dodero102,Reference Goedecke, Rae and Smuts104,Reference Kreider, Ferreira and Greenwood105) were deemed the moderate risk of bias and the remaining eighteen(Reference Chen, Lin and Huang23,Reference Gaullier, Halse and Høye45,Reference Larsen, Toubro and Gudmundsen48,Reference López-Plaza, Bermejo and Koester Weber49,Reference Norris, Collene and Asp55,Reference Sluijs, Plantinga and de Roos64,Reference Chang, Gan and Liao71,Reference Ebrahimi-Mameghani, Jamali and Mahdavi73Reference Ghobadi, Matin and Nemati75,Reference Rezvani, Montazeri and Baradaran84Reference Adams, Hsueh and Alford88,Reference Ribeiro, Pina and Dodero97,Reference Lopes, Silvestre and Silva101,Reference Attar-Bashi, Weisinger and Begg107) were at low risk of bias.

Meta-analysis results

Effects of conjugated linoleic acid supplementation on anthropometric measurements

Combining eighty-three effect sizes where CLA supplementation was compared with a placebo control revealed a significant lowering effect of CLA supplementation on BM (WMD: −0·34 kg, 95 % CI (−0·54, −0·15), P < 0·001). However, there was a significant between-study heterogeneity (I 2 = 42·7 %, P < 0·001) (Fig. 2(a)). The findings from subgroup analyses showed that CLA consumption was associated with decreased BM irrespective of the health condition of participants and follow-up length. In addition, BM was only reduced in overweight and obese individuals (as defined by BMI) and female and both sexes and in those who ingested 3 g/d or more of CLA. Weight-lowering effects of CLA were shown in both high- and low-quality studies but not in low-quality studies (Table 2).

Fig. 2. Forest plot detailing weighted mean difference and 95 % CI for the effect of CLA supplementation on: (a) body weight (kg); (b) BMI (kg/m2); (c) WC (cm); (d) FM (kg); (e) BFP (%); and (f) FFM (kg). CLA, conjugated linoleic acid; WC, waist circumference; FM, fat mas; BFP, body fat percentage; FFM, fat-free mass.

Table 2. Subgroup analyses of CLA supplementation on anthropometric indices and body composition

CLA, conjugated linoleic acid; WMD, weighted mean differences; WC, waist circumferenc; FM, fat mass; BFP, body fat percentage; FFM, fat-free mass.

Bold value: significant effect (p<0.05).

Eighty effect sizes from seventy-seven included RCT reported the effect of CLA supplementation on BMI and revealed a significant reduction in BMI (WMD: −0·15 kg/m2, 95 % CI (−0·24, −0·06), P = 0·001) albeit with a significant between-study heterogeneity (I 2 = 70·6 %, P < 0·001) (Fig. 2(b)). Subgroup analyses demonstrated BMI values were significantly reduced following CLA supplementation regardless of participant health status and follow-up duration. As with BM, BMI was only reduced in overweight and obese individuals and those who ingested 3 g/d or more CLA. However, BMI only decreased in high-quality studies subgroups (Table 2).

Overall, thirty-nine arms of included clinical trials investigated the effect of CLA supplementation on WC, and pooled effect size showed a significant reduction in WC (WMD: −0·67 cm, 95 % CI (−1·10, −0·23), P = 0·002) with significant between-study heterogeneity (I 2 = 76·0 %, P < 0·001) (Fig. 2(c)). Subgroup analyses revealed that CLA supplementation resulted in a significant reduction in WC among healthy participants, in those with baseline BMI > 30 kg/m2, upon supplementing with 3 g/d or more of CLA, and in cases where follow-up length was less than 12 weeks. Regarding the quality of studies, CLA was the cause of the WC decrease in moderate-quality studies (Table 2).

Effects of conjugated linoleic acid supplementation on body composition indicators

Meta-analysis of forty-nine effect sizes revealed a significant change in FM values after CLA intervention (WMD: −0·46 kg, 95 % CI (−0·68, −0·23), P < 0·001) despite a significant heterogeneity between studies (I 2 = 51·6 %, P < 0·001) (Fig. 2(d)). The findings of the subgroup analyses showed that CLA consumption reduced FM regardless of the intervention dosage and duration. However, CLA supplementation was associated with decreased FM only in healthy individuals and those with a baseline BMI categorised as overweight or obese. Furthermore, FM decreased in low- and moderate-quality subgroups (Table 2).

A total of forty-three effect sizes (835 cases and 808 controls) investigated the effect of CLA supplementation on BFP. Pooled data analysis indicated that BFP was significantly reduced following CLA supplementation compared with placebo (WMD: −0·76 %, 95 % CI (−1·08, −0·44), P < 0·001) albeit with a significant degree of heterogeneity between RCT (I 2 = 66·6 %, P < 0·001) (Fig. 2(e)). Subgroup analyses revealed that CLA supplementation significantly reduced BFP irrespective of participants’ health condition, baseline BMI values, and intervention dosages and duration. BFP-lowering effects of CLA are only seen in low-quality studies (Table 2).

Forty-five effect sizes (975 cases and 939 controls) were assessed for the effect of CLA supplementation on FFM. Meta-analysis indicated that CLA supplementation significantly increased FFM values in study participants (WMD: 0·27 kg, 95 % CI (0·09, 0·45), P = 0·003). A significant between-studies degree of heterogeneity was observed (I 2 = 47·6 %, P < 0·001) (Fig. 2(f)). Findings from subgroup analyses showed that CLA consumption was associated with increased FFM in healthy participants and in those with normal baseline BMI values upon supplementing with 3 g/d or more of CLA and when the trial duration was 12 weeks or more. Finally, FFM increases in high- and low-quality studies (Table 2).

Publication bias

There was no evidence of publication bias in RCT examining the effect of CLA supplementation for all outcomes, including BM (P = 0·142 Egger’s test), BMI (P = 0·201 Egger’s test), WC (P = 0·107 Egger’s test), FM (P = 0·055 Egger’s test), BFP (P = 0·059 Egger’s test) and FFM (P = 0·601 Egger’s test). Funnel plots further indicated no evidence of asymmetry for the effects of CLA consumption on each outcome analysed in this meta-analysis (online Supplementary 2).

Dose–response and meta-regression analyses

Dose–response analyses showed that CLA supplementation significantly altered BFP based on the intervention duration (r = −1·41, P-non-linearity = 0·04) in a non-linear manner. No other significant non-linear dose–response associations were observed for the remaining outcomes (online Supplementary 3 and 4). A meta-regression analysis was performed to assess the presence of any correlation between intervention duration (weeks) and dose of CLA supplementation with BM, BMI, WC, FM, BFP and FFM values. However, the meta-regression results demonstrated no significant linear relationship between changes in BM, BMI, WC, FM, BFP and FFM with the dose and duration of the intervention (online Supplementary 5 and 6).

Grading of evidence

An evaluation of the quality of evidence using the GRADE approach is presented in Table 3. For BM and FFM, the quality of evidence was high since included RCT had a low to moderate risk of bias with low statistical and clinical heterogeneity and narrow CI. Moreover, moderate quality of evidence was detected for BMI, WC, FM and BFP because of existing very serious limitations for inconsistency (I 2 = 69·5 %, I 2 = 75·0 %, I 2 = 51·8 %, and I 2 = 66·6 % for heterogeneity, respectively).

Table 3. GRADE profile of CLA supplementation for on anthropometric indices and body composition

GRADE, Grading of Recommendations Assessment, Development, and Evaluation; CLA, conjugated linoleic acid; WC, waist circumference; FM, fat mass; BFP, body fat percentage; FFM, fat-free mass.

* There is significant heterogeneity for BMI (I 2 = 70·6 %), WC (I 2 = 76·0 %), FM (I 2 = 51·6 %) and BFP (I 2 = 67·2 %).

Sensitivity analysis

Sensitivity analysis revealed that no particular RCT significantly influenced outcomes (BM, BMI, WC, FM, BFP and FFM) compared with others in a given data set (pooled effect).

Discussion

This meta-analysis showed that CLA supplementation decreased BM, BMI, WC, FM and BFP and increased FFM. It should be noted that the weight-loss properties of CLA were small and may not reach clinical importance. Based on subgroup analysis, CLA supplementation reduced BM and BMI only in overweight/obese individuals and those consuming more than 3 g/d of CLA. Moreover, CLA supplementation decreased WC in obese participants, dose ≥ 3 g/d and duration < 12 weeks. Regarding body composition indices, CLA supplementation only reduced FM in overweight/obese and in healthy participants. CLA intake as a dietary supplement increased FFM in healthy participants, normal BMI, dose ≥ 3 g/d and duration ≥ 12 weeks. Body composition improvement seems to be only statistically significant in females, not men. Meanwhile, a subgroup based on the quality of studies showed that high-quality studies failed to show the fat loss effects of CLA supplementation. However, high-quality studies showed a small but significant decrease in BM and BMI and an increase in FFM. The time–response model revealed that the optimal duration of CLA supplementation for reducing BFP was around 6 to 7 weeks. Variability in body fat distribution and susceptibility to obesity may explain this study’s failure to show a similar dose–response relationship of CLA supplementation with BFP, BM, BMI, WC, FM and FFM.

Numerous mechanisms of action in regulating body anthropometrics and composition following ingestion of CLA have been suggested, resulting from animal and human studies. For example, CLA seems to enhance fat mobilisation and oxidation, reduce the size of adipocytes, regulate lipolysis by adipocytes, increase apoptosis in preadipocytes and adipocytes, reduce adipocyte differentiation through interaction with PPAR-γ, and modulate cytokines-/adipokines-associated mechanisms(Reference Chen and Park108). Despite promising results in animal studies regarding an inverse relationship between CLA and obesity, evidence in humans supporting the role of CLA in reducing BM and improving repartitioning of body fat and FFM is limited.

As noted, early meta-analytic work by Whigham et al. (2007), having focused on the effects of CLA supplementation on body composition in the general adult population, indicated that by pooling effect estimates of eighteen RCT, CLA ingestion promoted moderate alterations in BF(Reference Whigham, Watras and Schoeller34). Moreover, Schoeller et al. (2009) conducted a meta-analytic study focused on eighteen trials, illustrating a small increase in FFM following CLA treatment(Reference Schoeller, Watras and Whigham35). Subsequent meta-analyses have focused on specific population outcomes, such as those who are overweight and/or obese, individuals with metabolic syndrome and postmenopausal women. For instance, Onakpoya et al. (2012) performed a meta-analysis on a select number of studies (n 7) and reported that consuming CLA for more than 6 months resulted in small yet significant reductions in BM, BMI and FM, along with no change in WC in overweight and obese and individuals(Reference Onakpoya, Posadzki and Watson32). Namazi et al. study (2019), working with thirteen trials utilising overweight and obese participants, showed that CLA slightly reduced BM, BMI, and FM and slightly increased lean body mass. Yet, these authors similarly reported no influence of CLA on WC measurements(Reference Namazi, Irandoost and Larijani31). A meta-analysis by Kim et al. (2016) conducted on nine RCT in metabolic syndrome patients showed BM and BMI improvements following CLA consumption. However, neither body composition nor WC was considered, thus limiting any direct comparisons between body composition and anthropometric alterations(Reference Kim, Lim and Lee33). Lastly, a recent meta-analysis (n 8) involving female participants performed by Hamdallah et al. (2020) illustrated that consuming CLA for between 6 and 16 weeks had moderate effects on BM, BMI and total body fat, particularly in those classified as overweight/obese and postmenopausal status(Reference Hamdallah, Ireland and Williams109).

In contrast with the meta-analyses mentioned above, the present study’s findings analysing an accumulation of seventy RCT demonstrated a small but significant efficacy for CLA supplementation to reduce WC. Moreover, some previous studies revealed that CLA could be a moderate anti-obesity agent without generating clinically relevant effects. This effect owes to CLA’s relatively limited reductions in BM (upwards of 5 %) and FM (approaching 8 %), as noted in prior investigations(Reference Chen and Park108,Reference Ryan and Yockey110) . Further, CLA administration might aid in targeted FM reduction (e.g. central abdominal fat pattern) rather than a more evenly distributed reduction of whole-body fat. Such modest effects of CLA in addressing obesity may also be advantageous when the risk of weight gain is heightened at particular times of the year (e.g. social occasions, holidays, etc.).

It should be noted that RCT in this study frequently used various types of vegetable oils as a placebo, including sunflower, olive, soyabean, paraffin, rapeseed and safflower. These oils are rich in MUFA and PUFA, like oleic acid, linoleic acid and α-linolenic acid. Biohydrogenation of linoleic acid into CLA may occur through the bacteria in the digestive tract and via the mediation of vaccenic acid. While it is assumed that these oils have supplementary or complementary effects which can influence human health(Reference Benjamin, Prakasan and Sreedharan111), overall effect size differences between CLA supplementation v. placebo may be muted in certain RCT, and care should be taken in future investigations to avoid such confounding variables. It is also worth noting that type of CLA supplement (isomer or mixture) as well varies in RCT where the trans-10 and cis-12 isomers of CLA are suggested to induce catabolic effects, including enhanced lipolysis and fat oxidation, while cis-9 and trans-11 are considered anabolic agent(Reference MacRedmond, Singhera and Attridge94).

Furthermore, applying different body composition measurement methodologies in clinical trials (e.g. bioelectrical impedance analysis v. dual-energy X-ray absorptiometry v. skinfold calipers) may influence the interpretation and accuracy of results. To alleviate such concerns, further clinical trials comparing CLA supplement types and selecting gold standard body composition methodologies should be undertaken.

While rare, a few RCT analysed in this meta-analysis reported complications during or following CLA intervention, amongst which gastrointestinal disorders were the most common. However, such unwanted side effects were not serious in the CLA dosage range reported in RCT, and generally, CLA appears to be safe and well tolerated.

Of other note, CLA taken with other supplements, dietary restriction and increased physical activity may further promote the correction of anthropometric indices and body composition in obese individuals(Reference Akkila112). For example, combining CLA with γ-oryzanol significantly reduced body fat in overweight Korean female participants(Reference Kim, Kim and Ha82). Another investigation on well-trained young adults indicated that CLA along with creatine and whey protein consumption enhanced strength gains and lean mass following heavy resistance training(Reference Cornish, Candow and Jantz113). Therefore, CLA intake and other weight-reducing or body composition-modulating treatments may provide additional benefits.

Although there is evidence outlining the small but significant effects of CLA supplementations on body composition, little is known about the impact of gender differences on body composition changes induced following CLA consumption. The gender-specific effect of different dietary interventions is important because it is generally more difficult for females to lose BM(Reference Williams, Wood and Collins114). Females are also likely to lose less BM than males during a dietary intervention(Reference Williams, Wood and Collins114), although they are more likely to adopt and adhere to a diet initially(Reference Kashubeck-West, Mintz and Weigold115). Although the findings of the gender differences in body composition changes induced by CLA supplementation in humans are limited, our study showed that CLA supplementation may be more beneficial in women than men. Further studies are needed to evaluate the gender-specific effects of CLA supplementation on body composition.

Strengths of this meta-analysis include a relatively large number of RCT containing no observable publication bias, suggesting overestimation of the relationship between CLA and body composition indicators and/or anthropometric measurements were avoided. Moreover, findings from sensitivity analyses support the robustness of the results. Finally, the quality of evidence was moderate to high. Limitations that should be acknowledged include identifying the sources of heterogeneity for BFP needed to be elucidated, and individuals with varying degrees of health status and other characteristics were pooled for overall effect size analyses, thus contributing to a rather heterogeneous sample. Heterogeneity was encountered, perhaps due to various regimens, doses, types, duration, centre settings and populations enrolled. Significant heterogeneity is a serious limitation and should be included because it may significantly undermine the validity of the result. Subgroup analyses were performed to find probable sources of heterogeneity based on the duration of studies, intervention dosage, participants’ health condition, obesity status and sex. However, significant heterogeneity in all included variables remains a main limitation in our findings. Moreover, the varying risk of bias in the pool of studies is another main limitation of our analysis. Although we conducted a subgroup analysis based on the quality of studies to minimise the limitation, another drawback is the devices used for body composition analysis in the included studies. Different body composition assessment methods do not always similarly reflect changes in body composition associated with weight loss. Finally, the present study has not been registered in the PROSPERO; this could also be considered a limitation.

In conclusion, CLA supplementation significantly, albeit mildly, reduces obesity markers, including BM, BMI, WC and FM, while enhancing FFM in an adult population. More specifically, anthropometric measures (BM, BMI and WC) improved following 3 g/d or more of CLA regardless of intervention duration (except for WC, which favoured shorter dosage durations of <12 weeks). In contrast, body composition alterations (FM, FFM and BFP) improved regardless of intervention dosage or duration (except for FFM, which favoured CLA dosages 3 g/d or more and longer duration trials lasting >12 weeks). Certain additional participant characteristics such as being overweight/obese (BM, BMI, WC and FM), noted as having healthy status (FM and FFM) and having normal BMI (FFM) further delineated the significance of overall effect size results. It should be noted that the data from high-quality studies failed to show the body fat-lowering properties of CLA. Also, both overall effects and high-quality studies showed that CLA supplementation resulted in weight loss. However, it should be noted that the weight-loss properties of CLA were small and may not reach clinical importance. It has been mentioned that the minimal clinically important difference is classified as clinically important and is considered the smallest effect required to produce clinically important results(Reference Setayesh, Ashtary-Larky and Clark116). The data for minimal clinically important difference regarding body composition are limited; however, a wide range of studies have confirmed that the risk of metabolic disorders could be decreased whenever they saw reductions of 5 % of initial weight(Reference Ashtary-Larky, Ghanavati and Lamuchi-Deli117,Reference Nascimento, Amorim and Alves118) . Warkentin et al. showed that weight reductions to achieve minimal clinically important difference for most health-related quality-of-life instruments are markedly higher than the conventional threshold of 5 % to 10 %. Future investigations should also determine the best combination of CLA with other anti-obesity agents to promote additional benefits of health-related BM parameters. Finally, to improve the continuity of results, the composition of fatty acids in a placebo should be carefully considered when investigating the effects of CLA in various populations.

Acknowledgements

This research received no specific grant from any funding agency, commercial or not-for-profit sectors.

O. A. contributed to the conception and design of the study; G. S. and M. Z. contributed to data extraction; S. S. and R. B. screened articles for inclusion criteria; O. A. contributed to data analysis, A. W., M. N., S. R., D. A. L. and K. N. contributed in manuscript drafting; O. A. and M. N. supervised the study. All authors approved the final version of the manuscript.

The authors declared that there is no conflict of interest.

Supplementary material

For supplementary material/s referred to in this article, please visit https://doi.org/10.1017/S0007114523001861

References

Orukwowu, U (2022) Epidemiology of adult obesity, measurements, global prevalence and risk factors. IPS Intell Multidiscip J 1, 16.Google Scholar
Hutchesson, MJ, Gough, C, Müller, AM, et al. (2021) eHealth interventions targeting nutrition, physical activity, sedentary behavior, or obesity in adults: a scoping review of systematic reviews. Obes Rev 22, e13295.10.1111/obr.13295CrossRefGoogle ScholarPubMed
Wang, YC, McPherson, K, Marsh, T, et al. (2011) Health and economic burden of the projected obesity trends in the USA and the UK. Lancet 378, 815825.CrossRefGoogle ScholarPubMed
Ashtary-Larky, D, Bagheri, R, Bavi, H, et al. (2022) Ketogenic diets, physical activity and body composition: a review. Br J Nutr 127, 18981920.10.1017/S0007114521002609CrossRefGoogle ScholarPubMed
Ashtary-Larky, D, Bagheri, R, Tinsley, GM, et al. (2021) Effects of intermittent fasting combined with resistance training on body composition: a systematic review and meta-analysis. Physiol Behav 237, 113453.CrossRefGoogle ScholarPubMed
Ashtary-Larky, D, Bagheri, R, Abbasnezhad, A, et al. (2020) Effects of gradual weight loss v. rapid weight loss on body composition and RMR: a systematic review and meta-analysis. Br J Nutr 124, 11211132.10.1017/S000711452000224XCrossRefGoogle ScholarPubMed
Asbaghi, O, Naeini, F, Ashtary-Larky, D, et al. (2021) Effects of chromium supplementation on blood pressure, body mass index, liver function enzymes and malondialdehyde in patients with type 2 diabetes: a systematic review and dose-response meta-analysis of randomized controlled trials. Complement Ther Med 60, 102755.CrossRefGoogle ScholarPubMed
Naseri, K, Saadati, S, Yari, Z, et al. (2022) Beneficial effects of probiotic and synbiotic supplementation on some cardiovascular risk factors among individuals with prediabetes and type 2 diabetes mellitus: a grade-assessed systematic review, meta-analysis, and meta-regression of randomized clinical trials. Pharmacol Res 182, 106288.10.1016/j.phrs.2022.106288CrossRefGoogle ScholarPubMed
Bagheri, R, Rashidlamir, A, Ashtary-Larky, D, et al. (2020) Does green tea extract enhance the anti-inflammatory effects of exercise on fat loss? Br J Clin Pharmacol 86, 753762.CrossRefGoogle ScholarPubMed
Bagheri, R, Negaresh, R, Motevalli, MS, et al. (2022) Spirulina supplementation during gradual weight loss in competitive wrestlers. Br J Nutr 127, 248256.10.1017/S000711452100091XCrossRefGoogle ScholarPubMed
Chang, H, Gan, W, Liao, X, et al. (2020) Conjugated linoleic acid supplements preserve muscle in high-body-fat adults: a double-blind, randomized, placebo trial. Nutr Metab Cardiovasc Dis 30, 17771784.10.1016/j.numecd.2020.05.029CrossRefGoogle ScholarPubMed
Mądry, E, Malesza, IJ, Subramaniapillai, M, et al. (2020) Body fat changes and liver safety in obese and overweight women supplemented with conjugated linoleic acid: a 12-week randomised, double-blind, placebo-controlled trial. Nutrients 12, 1811.10.3390/nu12061811CrossRefGoogle ScholarPubMed
Basak, S & Duttaroy, AK (2020) Conjugated linoleic acid and its beneficial effects in obesity, cardiovascular disease, and cancer. Nutrients 12, 1913.10.3390/nu12071913CrossRefGoogle ScholarPubMed
den Hartigh, LJ, Wang, S, Goodspeed, L, et al. (2017) Metabolically distinct weight loss by 10, 12 CLA and caloric restriction highlight the importance of subcutaneous white adipose tissue for glucose homeostasis in mice. PLOS ONE 12, e0172912.CrossRefGoogle ScholarPubMed
Koba, K & Yanagita, T (2014) Health benefits of conjugated linoleic acid (CLA). Obes Res Clin Pract 8, e525e532.10.1016/j.orcp.2013.10.001CrossRefGoogle ScholarPubMed
O’Reilly, ME, Lenighan, YM, Dillon, E, et al. (2020) Conjugated linoleic acid and α linolenic acid improve cholesterol homeostasis in obesity by modulating distinct hepatic protein pathways. Mol Nutr Food Res 64, 1900599.CrossRefGoogle ScholarPubMed
Asbaghi, O, Ashtary-Larky, D, Naseri, K, et al. (2022) The effects of conjugated linoleic acid supplementation on lipid profile in adults: a systematic review and dose-response meta-analysis. Front Nutr 9, 953012.CrossRefGoogle ScholarPubMed
Haghighat, N, Shimi, G, Shiraseb, F, et al. (2022) The effects of conjugated linoleic acid supplementation on liver function enzymes and malondialdehyde in adults: a GRADE-assessed systematic review and dose-response meta-analysis. Pharmacol Res 186, 106518.CrossRefGoogle ScholarPubMed
Ashwell, MS, Ceddia, RP, House, RL, et al. (2010) Trans-10, cis-12-conjugated linoleic acid alters hepatic gene expression in a polygenic obese line of mice displaying hepatic lipidosis. J Nutr Biochem 21, 848855.10.1016/j.jnutbio.2009.06.013CrossRefGoogle Scholar
Churruca, I, Fernández-Quintela, A, Zabala, A, et al. (2007) The effect of trans-10, cis-12 conjugated linoleic acid on lipogenesis is tissue dependent in hamsters. Genes Nutr 2, 121123.10.1007/s12263-007-0031-8CrossRefGoogle ScholarPubMed
Sato, K, Shinohara, N, Honma, T, et al. (2011) The change in conjugated linoleic acid concentration in blood of Japanese fed a conjugated linoleic acid diet. J Nutr Sci Vitaminol 57, 364371.10.3177/jnsv.57.364CrossRefGoogle ScholarPubMed
Turpeinen, AM, Mutanen, M, Aro, A, et al. (2002) Bioconversion of vaccenic acid to conjugated linoleic acid in humans. Am J Clin Nutr 76, 504510.CrossRefGoogle ScholarPubMed
Chen, SC, Lin, YH, Huang, HP, et al. (2012) Effect of conjugated linoleic acid supplementation on weight loss and body fat composition in a Chinese population. Nutrition 28, 559565.CrossRefGoogle ScholarPubMed
Kanter, JE, Goodspeed, L, Wang, S, et al. (2018) 10,12 Conjugated linoleic acid-driven weight loss is protective against atherosclerosis in mice and is associated with alternative macrophage enrichment in perivascular adipose tissue. Nutrients 10, 1416.CrossRefGoogle Scholar
Mądry, E, Chudzicka-Strugała, I, Grabańska-Martyńska, K, et al. (2016) Twelve weeks CLA supplementation decreases the hip circumference in overweight and obese women. A double-blind, randomized, placebo-controlled trial. Acta Sci Pol Technol Aliment 15, 107113.10.17306/J.AFS.2016.1.11CrossRefGoogle ScholarPubMed
Baddini Feitoza, A, Fernandes Pereira, A, Ferreira da Costa, N, et al. (2009) Conjugated linoleic acid (CLA): effects on modulating body composition and lipid profile. Nutr Hosp 24, 422428.Google ScholarPubMed
Brown, JM & McIntosh, MK (2003) Conjugated linoleic acid in humans: regulation of adiposity and insulin sensitivity. J Nutr 133, 30413046.10.1093/jn/133.10.3041CrossRefGoogle ScholarPubMed
Joseph, L, Wasir, JS, Misra, A, et al. (2011) Appropriate values of adiposity and lean body mass indices to detect cardiovascular risk factors in Asian Indians. Diabetes Technol Ther 13, 899906.CrossRefGoogle ScholarPubMed
Pi-Sunyer, FX (2000) Obesity: criteria and classification. Proc Nutr Soc 59, 505509.CrossRefGoogle ScholarPubMed
Abedi, R, Aref-Hosseini, S-R, Khoshbaten, M, et al. (2018) The Effect of conjugated linoleic acid (CLA) on inflammatory factors in non-alcoholic fatty liver disease (NAFLD): a randomized controlled clinical trial. Progr Nutr 20, 173181.Google Scholar
Namazi, N, Irandoost, P, Larijani, B, et al. (2019) The effects of supplementation with conjugated linoleic acid on anthropometric indices and body composition in overweight and obese subjects: a systematic review and meta-analysis. Crit Rev Food Sci Nutr 59, 27202733.CrossRefGoogle ScholarPubMed
Onakpoya, IJ, Posadzki, PP, Watson, LK, et al. (2012) The efficacy of long-term conjugated linoleic acid (CLA) supplementation on body composition in overweight and obese individuals: a systematic review and meta-analysis of randomized clinical trials. Eur J Nutr 51, 127134.CrossRefGoogle ScholarPubMed
Kim, B, Lim, HR, Lee, H, et al. (2016) The effects of conjugated linoleic acid (CLA) on metabolic syndrome patients: a systematic review and meta-analysis. J Funct Foods 25, 588598.10.1016/j.jff.2016.07.010CrossRefGoogle Scholar
Whigham, LD, Watras, AC & Schoeller, DA (2007) Efficacy of conjugated linoleic acid for reducing fat mass: a meta-analysis in humans. Am J Clin Nutr 85, 12031211.10.1093/ajcn/85.5.1203CrossRefGoogle Scholar
Schoeller, DA, Watras, AC & Whigham, LD (2009) A meta-analysis of the effects of conjugated linoleic acid on fat-free mass in humans. Appl Physiol Nutr Metab 34, 975978.10.1139/H09-080CrossRefGoogle ScholarPubMed
Moher, D, Shamseer, L, Clarke, M, et al. (2015) Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Syst Rev 4, 1.CrossRefGoogle ScholarPubMed
Higgins, JP, Thomas, J, Chandler, J, et al. (2019) Cochrane Handbook for Systematic Reviews of Interventions. Chichester: John Wiley & Sons.10.1002/9781119536604CrossRefGoogle Scholar
Guyatt, GH, Oxman, AD, Vist, GE, et al. (2008) GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ 336, 924926.10.1136/bmj.39489.470347.ADCrossRefGoogle ScholarPubMed
Higgins, Julian PT & Sally Green, eds. (2008). Cochrane handbook for systematic reviews of interventions: S38.10.1002/9780470712184CrossRefGoogle Scholar
Egger, M, Davey Smith, G, Schneider, M, et al. (1997) Bias in meta-analysis detected by a simple, graphical test. BMJ 315, 629634.CrossRefGoogle ScholarPubMed
Berven, G, Bye, A, Hals, O, et al. (2000) Safety of conjugated linoleic acid (CLA) in overweight or obese human volunteers. Eur J Lipid Sci Technol 102, 455462.10.1002/1438-9312(200008)102:7<455::AID-EJLT455>3.0.CO;2-V3.0.CO;2-V>CrossRefGoogle Scholar
Blankson, H, Stakkestad, JA, Fagertun, H, et al. (2000) Conjugated linoleic acid reduces body fat mass in overweight and obese humans. J Nutr 130, 29432948.Google ScholarPubMed
Gaullier, JM, Halse, J, Høivik, HO, et al. (2007) Six months supplementation with conjugated linoleic acid induces regional-specific fat mass decreases in overweight and obese. Br J Nutr 97, 550560.CrossRefGoogle ScholarPubMed
Gaullier, JM, Halse, J, Høye, K, et al. (2004) Conjugated linoleic acid supplementation for 1 year reduces body fat mass in healthy overweight humans. Am J Clin Nutr 79, 11181125.CrossRefGoogle Scholar
Gaullier, JM, Halse, J, Høye, K, et al. (2005) Supplementation with conjugated linoleic acid for 24 months is well tolerated by and reduces body fat mass in healthy, overweight humans. J Nutr 135, 778784.CrossRefGoogle ScholarPubMed
Kamphuis, MM, Lejeune, MP, Saris, WH, et al. (2003) The effect of conjugated linoleic acid supplementation after weight loss on body weight regain, body composition, and resting metabolic rate in overweight subjects. Int J Obes Relat Metab Disord 27, 840847.10.1038/sj.ijo.0802304CrossRefGoogle ScholarPubMed
Kamphuis, MM, Lejeune, MP, Saris, WH, et al. (2003) Effect of conjugated linoleic acid supplementation after weight loss on appetite and food intake in overweight subjects. Eur J Clin Nutr 57, 12681274.10.1038/sj.ejcn.1601684CrossRefGoogle ScholarPubMed
Larsen, TM, Toubro, S, Gudmundsen, O, et al. (2006) Conjugated linoleic acid supplementation for 1 year does not prevent weight or body fat regain. Am J Clin Nutr 83, 606612.CrossRefGoogle ScholarPubMed
López-Plaza, B, Bermejo, LM, Koester Weber, T, et al. (2013) Effects of milk supplementation with conjugated linoleic acid on weight control and body composition in healthy overweight people. Nutr Hosp 28, 20902098.Google ScholarPubMed
Mądry, E, Malesza, IJ, Subramaniapillai, M, et al. (2020) Body fat changes and liver safety in obese and overweight women supplemented with conjugated linoleic acid: a 12-week randomised, double-blind, placebo-controlled trial. Nutrients 12, 1811.CrossRefGoogle ScholarPubMed
Malpuech-Brugère, C, Verboeket-Van de Venne, WP, Mensink, RP, et al. (2004) Effects of two conjugated linoleic acid isomers on body fat mass in overweight humans. Obes Res 12, 591598.10.1038/oby.2004.68CrossRefGoogle ScholarPubMed
Mougios, V, Matsakas, A, Petridou, A, et al. (2001) Effect of supplementation with conjugated linoleic acid on human serum lipids and body fat. J Nutr Biochem 12, 585594.10.1016/S0955-2863(01)00177-2CrossRefGoogle ScholarPubMed
Nazare, JA, de la Perrière, AB, Bonnet, F, et al. (2007) Daily intake of conjugated linoleic acid-enriched yoghurts: effects on energy metabolism and adipose tissue gene expression in healthy subjects. Br J Nutr 97, 273280.10.1017/S0007114507191911CrossRefGoogle ScholarPubMed
Noone, EJ, Roche, HM, Nugent, AP, et al. (2002) The effect of dietary supplementation using isomeric blends of conjugated linoleic acid on lipid metabolism in healthy human subjects. Br J Nutr 88, 243251.CrossRefGoogle ScholarPubMed
Norris, LE, Collene, AL, Asp, ML, et al. (2009) Comparison of dietary conjugated linoleic acid with safflower oil on body composition in obese postmenopausal women with type 2 diabetes mellitus. Am J Clin Nutr 90, 468476.CrossRefGoogle ScholarPubMed
Nugent, AP, Roche, HM, Noone, EJ, et al. (2005) The effects of conjugated linoleic acid supplementation on immune function in healthy volunteers. Eur J Clin Nutr 59, 742750.CrossRefGoogle ScholarPubMed
Pfeuffer, M, Fielitz, K, Laue, C, et al. (2011) CLA does not impair endothelial function and decreases body weight as compared with safflower oil in overweight and obese male subjects. J Am Coll Nutr 30, 1928.CrossRefGoogle Scholar
Raff, M, Tholstrup, T, Basu, S, et al. (2008) A diet rich in conjugated linoleic acid and butter increases lipid peroxidation but does not affect atherosclerotic, inflammatory, or diabetic risk markers in healthy young men. J Nutr 138, 509514.CrossRefGoogle ScholarPubMed
Risérus, U, Arner, P, Brismar, K, et al. (2002) Treatment with dietary trans10cis12 conjugated linoleic acid causes isomer-specific insulin resistance in obese men with the metabolic syndrome. Diabetes Care 25, 15161521.10.2337/diacare.25.9.1516CrossRefGoogle ScholarPubMed
Risérus, U, Berglund, L & Vessby, B (2001) Conjugated linoleic acid (CLA) reduced abdominal adipose tissue in obese middle-aged men with signs of the metabolic syndrome: a randomised controlled trial. Int J Obes Relat Metab Disord 25, 11291135.CrossRefGoogle ScholarPubMed
Risérus, U, Vessby, B, Arner, P, et al. (2004) Supplementation with trans10cis12-conjugated linoleic acid induces hyperproinsulinaemia in obese men: close association with impaired insulin sensitivity. Diabetologia 47, 10161019.CrossRefGoogle ScholarPubMed
Risérus, U, Vessby, B, Arnlöv, J, et al. (2004) Effects of cis-9,trans-11 conjugated linoleic acid supplementation on insulin sensitivity, lipid peroxidation, and proinflammatory markers in obese men. Am J Clin Nutr 80, 279283.CrossRefGoogle ScholarPubMed
Rubin, D, Herrmann, J, Much, D, et al. (2012) Influence of different CLA isomers on insulin resistance and adipocytokines in pre-diabetic, middle-aged men with PPARγ2 Pro12Ala polymorphism. Genes Nutr 7, 499509.CrossRefGoogle ScholarPubMed
Sluijs, I, Plantinga, Y, de Roos, B, et al. (2010) Dietary supplementation with cis-9,trans-11 conjugated linoleic acid and aortic stiffness in overweight and obese adults. Am J Clin Nutr 91, 175183.CrossRefGoogle ScholarPubMed
Sneddon, AA, Tsofliou, F, Fyfe, CL, et al. (2008) Effect of a conjugated linoleic acid and n-3 fatty acid mixture on body composition and adiponectin. Obesity 16, 10191024.CrossRefGoogle ScholarPubMed
Steck, SE, Chalecki, AM, Miller, P, et al. (2007) Conjugated linoleic acid supplementation for 12 weeks increases lean body mass in obese humans. J Nutr 137, 11881193.CrossRefGoogle ScholarPubMed
Taylor, JS, Williams, SR, Rhys, R, et al. (2006) Conjugated linoleic acid impairs endothelial function. Arterioscler Thromb Vasc Biol 26, 307312.CrossRefGoogle ScholarPubMed
Thom, E, Wadstein, J & Gudmundsen, O (2001) Conjugated linoleic acid reduces body fat in healthy exercising humans. J Int Med Res 29, 392396.10.1177/147323000102900503CrossRefGoogle ScholarPubMed
Aryaeian, N, Shahram, F, Djalali, M, et al. (2008) Effect of conjugated linoleic acid, vitamin E and their combination on lipid profiles and blood pressure of Iranian adults with active rheumatoid arthritis. Vasc Health Risk Manag 4, 14231432.10.2147/VHRM.S3822CrossRefGoogle ScholarPubMed
Bulut, S, Bodur, E, Colak, R, et al. (2013) Effects of conjugated linoleic acid supplementation and exercise on post-heparin lipoprotein lipase, butyrylcholinesterase, blood lipid profile and glucose metabolism in young men. Chem Biol Interact 203, 323329.CrossRefGoogle ScholarPubMed
Chang, H, Gan, W, Liao, X, et al. (2020) Conjugated linoleic acid supplements preserve muscle in high-body-fat adults: a double-blind, randomized, placebo trial. Nutr Metab Cardiovasc Dis 30, 17771784.CrossRefGoogle ScholarPubMed
Colakoglu, S, Colakoglu, M, Taneli, F, et al. (2006) Cumulative effects of conjugated linoleic acid and exercise on endurance development, body composition, serum leptin and insulin levels. J Sports Med Phys Fitness 46, 570577.Google ScholarPubMed
Ebrahimi-Mameghani, M, Jamali, H, Mahdavi, R, et al. (2016) Conjugated linoleic acid improves glycemic response, lipid profile, and oxidative stress in obese patients with non-alcoholic fatty liver disease: a randomized controlled clinical trial. Croat Med J 57, 331342.10.3325/cmj.2016.57.331CrossRefGoogle ScholarPubMed
Esmaeili Shahmirzadi, F, Ghavamzadeh, S & Zamani, T (2019) The effect of conjugated linoleic acid supplementation on body composition, serum insulin and leptin in obese adults. Arch Iran Med 22, 255261.Google ScholarPubMed
Ghobadi, H, Matin, S, Nemati, A, et al. (2016) The effect of conjugated linoleic acid supplementation on the nutritional status of COPD patients. Int J Chron Obstruct Pulmon Dis 11, 27112720.CrossRefGoogle ScholarPubMed
Hassan Eftekhari, M, Aliasghari, F, Babaei-Beigi, MA, et al. (2013) Effect of conjugated linoleic acid and n-3 fatty acid supplementation on inflammatory and oxidative stress markers in atherosclerotic patients. ARYA Atheroscler 9, 311318.Google ScholarPubMed
Michishita, T, Kobayashi, S, Katsuya, T, et al. (2010) Evaluation of the antiobesity effects of an amino acid mixture and conjugated linoleic acid on exercising healthy overweight humans: a randomized, double-blind, placebo-controlled trial. J Int Med Res 38, 844859.CrossRefGoogle ScholarPubMed
Shadman, Z, Taleban, FA, Saadat, N, et al. (2013) Effect of conjugated linoleic acid and vitamin E on glycemic control, body composition, and inflammatory markers in overweight type2 diabetics. J Diabetes Metab Disord 12, 42.CrossRefGoogle ScholarPubMed
Tajmanesh, M, Aryaeian, N, Hosseini, M, et al. (2015) Conjugated linoleic acid supplementation has no impact on aerobic capacity of healthy young men. Lipids 50, 805809.CrossRefGoogle ScholarPubMed
Zhao, WS, Zhai, JJ, Wang, YH, et al. (2009) Conjugated linoleic acid supplementation enhances antihypertensive effect of ramipril in Chinese patients with obesity-related hypertension. Am J Hypertens 22, 680686.10.1038/ajh.2009.56CrossRefGoogle ScholarPubMed
Fouladi, H, Peng, LS & Mohaghehgi, A (2018) Effects of conjugated linoleic acid supplementation and exercise on body fat mass and blood lipid profiles among overweight Iranians. Mal J Nutr 24, 203213.Google Scholar
Kim, J-H, Kim, O-H, Ha, Y-L, et al. (2008) Supplementation of conjugated linoleic acid with γ-oryzanol for 12 weeks effectively reduces body fat in healthy overweight Korean women. Prev Nutr Food Sci 13, 146156.CrossRefGoogle Scholar
Park, E-J, Kim, J-M, Kim, K-T, et al. (2008) Conjugated linoleic acid (CLA) supplementation for 8 weeks reduces body weight in healthy overweight/obese Korean subjects. Food Sci Biotechnol 17, 12611264.Google Scholar
Rezvani, N, Montazeri, V, Baradaran, B, et al. (2018) Effects of conjugated fatty acid supplementation on central obesity and blood pressure in women with benign breast disease: a randomized controlled-clinical trial. Progr Nutr 20, 163172.Google Scholar
Son, S-J, Lee, J-E, Park, E-K, et al. (2009) The effects of conjugated linoleic acid and/or exercise on body weight and body composition in college women with high body fat mass. Korean J Food Sci Technol 41, 307312.Google Scholar
Tavakkoli Darestani, A, Hosseinpanah, F, Tahbaz, F, et al. (2010) Effects of conjugated linoleic acid supplementation on body composition and leptin concentration in post-menopausal women. Iran J Endocrinol Metab 12, 4859.Google Scholar
Zh, S, Rastmanesh, S & Hehayati, M (2011) Effect of conjugated linoleic acid on serum leptin, adiponectin and body composition in overweight type II diabetic patients. Trauma Mon 2011, 101107.Google Scholar
Adams, RE, Hsueh, A, Alford, B, et al. (2006) Conjugated linoleic acid supplementation does not reduce visceral adipose tissue in middle-aged men engaged in a resistance-training program. J Int Soc Sports Nutr 3, 2836.CrossRefGoogle ScholarPubMed
Brown, AW, Trenkle, AH & Beitz, DC (2011) Diets high in conjugated linoleic acid from pasture-fed cattle did not alter markers of health in young women. Nutr Res 31, 3341.CrossRefGoogle Scholar
Carvalho, RF, Uehara, SK & Rosa, G (2012) Microencapsulated conjugated linoleic acid associated with hypocaloric diet reduces body fat in sedentary women with metabolic syndrome. Vasc Health Risk Manag 8, 661667.CrossRefGoogle ScholarPubMed
Deguire, JR, Makarem, N, Vanstone, CA, et al. (2012) Conjugated linoleic acid is related to bone mineral density but does not affect parathyroid hormone in men. Nutr Res 32, 911920.CrossRefGoogle Scholar
Desroches, S, Chouinard, PY, Galibois, I, et al. (2005) Lack of effect of dietary conjugated linoleic acids naturally incorporated into butter on the lipid profile and body composition of overweight and obese men. Am J Clin Nutr 82, 309319.CrossRefGoogle ScholarPubMed
Joseph, SV, Jacques, H, Plourde, M, et al. (2011) Conjugated linoleic acid supplementation for 8 weeks does not affect body composition, lipid profile, or safety biomarkers in overweight, hyperlipidemic men. J Nutr 141, 12861291.10.3945/jn.110.135087CrossRefGoogle ScholarPubMed
MacRedmond, R, Singhera, G, Attridge, S, et al. (2010) Conjugated linoleic acid improves airway hyper-reactivity in overweight mild asthmatics. Clin Exp Allergy 40, 10711078.CrossRefGoogle ScholarPubMed
Medina, EA, Horn, WF, Keim, NL, et al. (2000) Conjugated linoleic acid supplementation in humans: effects on circulating leptin concentrations and appetite. Lipids 35, 783788.CrossRefGoogle ScholarPubMed
Pinkoski, C, Chilibeck, PD, Candow, DG, et al. (2006) The effects of conjugated linoleic acid supplementation during resistance training. Med Sci Sports Exerc 38, 339348.10.1249/01.mss.0000183860.42853.15CrossRefGoogle ScholarPubMed
Ribeiro, AS, Pina, FL, Dodero, SR, et al. (2016) Effect of conjugated linoleic acid associated with aerobic exercise on body fat and lipid profile in obese women: a randomized, double-blinded, and placebo-controlled trial. Int J Sport Nutr Exerc Metab 26, 135144.CrossRefGoogle ScholarPubMed
Venkatramanan, S, Joseph, SV, Chouinard, PY, et al. (2010) Milk enriched with conjugated linoleic acid fails to alter blood lipids or body composition in moderately overweight, borderline hyperlipidemic individuals. J Am Coll Nutr 29, 152159.10.1080/07315724.2010.10719829CrossRefGoogle ScholarPubMed
Watras, AC, Buchholz, AC, Close, RN, et al. (2007) The role of conjugated linoleic acid in reducing body fat and preventing holiday weight gain. Int J Obes 31, 481487.CrossRefGoogle ScholarPubMed
Zambell, KL, Keim, NL, Van Loan, MD, et al. (2000) Conjugated linoleic acid supplementation in humans: effects on body composition and energy expenditure. Lipids 35, 777782.CrossRefGoogle ScholarPubMed
Lopes, DCF, Silvestre, MPC, Silva, VDM, et al. (2013) Dietary supplementation of conjugated linoleic acid, added to a milk drink, in women. Asian J Sci Res 6, 679.10.3923/ajsr.2013.679.690CrossRefGoogle Scholar
Pina, FLC, Ribeiro, AS, Dodero, SR, et al. (2016) Conjugated linoleic acid supplementation does not maximize motor performance and abdominal and trunk fat loss induced by aerobic training in overweight women. Rev Nutr 29, 785795.CrossRefGoogle Scholar
Plourde, M (2011) Conjugated linoleic acid supplementation for 8 weeks fails to impact body composition, lipid profile, or safety parameters in overweight, hyperlipidemic men. J Nutr 141, 12861291.Google Scholar
Goedecke, JH, Rae, DE, Smuts, CM, et al. (2009) Conjugated linoleic acid isomers, t10c12 and c9t11, are differentially incorporated into adipose tissue and skeletal muscle in humans. Lipids 44, 983988.CrossRefGoogle ScholarPubMed
Kreider, RB, Ferreira, MP, Greenwood, M, et al. (2002) Effects of conjugated linoleic acid supplementation during resistance training on body composition, bone density, strength, and selected hematological markers. J Strength Cond Res 16, 325334.Google ScholarPubMed
Lambert, EV, Goedecke, JH, Bluett, K, et al. (2007) Conjugated linoleic acid versus high-oleic acid sunflower oil: effects on energy metabolism, glucose tolerance, blood lipids, appetite and body composition in regularly exercising individuals. Br J Nutr 97, 10011011.CrossRefGoogle ScholarPubMed
Attar-Bashi, NM, Weisinger, RS, Begg, DP, et al. (2007) Failure of conjugated linoleic acid supplementation to enhance biosynthesis of docosahexaenoic acid from α-linolenic acid in healthy human volunteers. Prostaglandins Leukot Essent Fatty Acids 76, 121130.CrossRefGoogle ScholarPubMed
Chen, PB & Park, Y (2019) Conjugated linoleic acid in human health: effects on weight control. Nutr Prev Treat Abdom Obes, 355382.Google Scholar
Hamdallah, H, Ireland, HE & Williams, J (2020) Conjugated linoleic acid (CLA) effect on body weight and body composition in women (systematic review and meta-analysis). Proc Nutr Soc 79, 262.CrossRefGoogle Scholar
Ryan, DH & Yockey, SR (2017) Weight loss and improvement in comorbidity: differences at 5 %, 10 %, 15 %, and over. Curr Obes Rep 6, 187194.CrossRefGoogle ScholarPubMed
Benjamin, S, Prakasan, P, Sreedharan, S, et al. (2015) Pros and cons of CLA consumption: an insight from clinical evidences. Nutr Metab 12, 121.CrossRefGoogle ScholarPubMed
Akkila, SS (2018) Conjugated linoleic acid accelerates weight loss and improves anthropometric measures in overweight young adult males during weight loss program. Obes Metab 15, 1924.CrossRefGoogle Scholar
Cornish, SM, Candow, DG, Jantz, NT, et al. (2009) Conjugated linoleic acid combined with creatine monohydrate and whey protein supplementation during strength training. Int J Sport Nutr Exerc Metab 19, 7996.CrossRefGoogle ScholarPubMed
Williams, RL, Wood, LG, Collins, CE, et al. (2015) Effectiveness of weight loss interventions – is there a difference between men and women: a systematic review. Obes Rev 16, 171186.CrossRefGoogle Scholar
Kashubeck-West, S, Mintz, LB & Weigold, I (2005) Separating the effects of gender and weight-loss desire on body satisfaction and disordered eating behavior. Sex Roles 53, 505518.CrossRefGoogle Scholar
Setayesh, L, Ashtary-Larky, D, Clark, CCT, et al. (2021) The effect of saffron supplementation on blood pressure in adults: a systematic review and dose-response meta-analysis of randomized controlled trials. Nutrients 13, 2736.CrossRefGoogle ScholarPubMed
Ashtary-Larky, D, Ghanavati, M, Lamuchi-Deli, N, et al. (2017) Rapid weight loss v. slow weight loss: which is more effective on body composition and metabolic risk factors? Int J Endocrinol Metab 15, e13249.Google Scholar
Nascimento, DDC, Amorim, DNP, Alves, VP, et al. (2022) Reply on “significant change for body composition data”. Osteoporos Sarcopenia 8, 132133.10.1016/j.afos.2022.09.004CrossRefGoogle Scholar
Figure 0

Fig. 1. Flow chart of study selection for inclusion trials in the systematic review.

Figure 1

Table 1. Characteristics of the included studies

Figure 2

Fig. 2. Forest plot detailing weighted mean difference and 95 % CI for the effect of CLA supplementation on: (a) body weight (kg); (b) BMI (kg/m2); (c) WC (cm); (d) FM (kg); (e) BFP (%); and (f) FFM (kg). CLA, conjugated linoleic acid; WC, waist circumference; FM, fat mas; BFP, body fat percentage; FFM, fat-free mass.

Figure 3

Table 2. Subgroup analyses of CLA supplementation on anthropometric indices and body composition

Figure 4

Table 3. GRADE profile of CLA supplementation for on anthropometric indices and body composition

Supplementary material: File

Asbaghi et al. supplementary material
Download undefined(File)
File 334.4 KB